You are on page 1of 9

South African Journal of Botany 130 (2020) 4553

Contents lists available at ScienceDirect

South African Journal of Botany


journal homepage: www.elsevier.com/locate/sajb

Evaluation of the antioxidant and anti-arthritic potential of Zingiber


officinale Rosc. by in vitro and in silico analysis
Selvakumar Murugesan, Meenakshi R. Venkateswaran, Sasidharan Jayabal,
Sureshkumar Periyasamy*
Department of Biotechnology, Anna University, BIT-Campus, Tiruchirappalli 620024, Tamil Nadu, India

A R T I C L E I N F O A B S T R A C T

Article History: Rheumatoid Arthritis (RA) is an autoimmune disorder that leads to joint and bone destruction. The available
Received 30 July 2019 anti-inflammatory chemo drugs are associated with adverse side-effects providing only temporary relief.
Revised 6 November 2019 Herbal medicine can act as an alternative source for RA treatment with improved efficacy, lesser toxicity and
Accepted 11 December 2019
side effects. An attempt was made to evaluate the antioxidant and anti-arthritic potential of ginger (Zingiber
Available online xxx
officinale Rosc.). Crude ginger extracts were prepared with different organic solvents using soxhlet approach.
Edited by L R 
arova The extracts were assessed for their anti-oxidant activity using DPPH, ABTS and nitric oxide radical inhibition
Keywords:
assays and anti-arthritic activity through membrane stabilization assay, anti-proteinase activity and protein
Zingiber officinale denaturation inhibition assays. The highest scavenging potential was exhibited by Z. officinale methanolic
Antioxidant extract (ZOME) as assessed by DPPH assay (86.26 § 0.97%), ABTS assay (91.04 § 0.96%) and nitric oxide assay
Anti rheumatic (86.72 § 1.51%). Similarly, the ZOME showed 84.72 § 1.38% of inhibition in membrane stabilization assay,
8-Gingerol 82.72 § 1.48% in anti-proteinase activity and 81.68 § 1.66% in protein denaturation inhibition capacity. GC-
Bioavailability MS analysis of ZOME showed 30 different phytoconstituents. The major ginger bioactive compounds were
In silico virtually docked with novel RA targets using PyRx and the pharmacokinetic properties like ADMET proper-
ties were evaluated. Among all, 8-Gingerol showed the highest covalent interaction along with suitable bind-
ing affinity to the RA targets of COX-II (7.2), TNF-a (4.7), MCSF (5.0), IL-1b (5.7) and MMP9 (6.7) kcal/
mol. 8-Gingerol can be a better drug candidate that must be further investigated for mechanistic studies to
verify its therapeutic potential in treating RA.
© 2019 Published by Elsevier B.V. on behalf of SAAB.

1. Introduction are the most potential target proteins for RA treatment at chronic
inflammatory conditions. The chemodrugs used for the treatment of
Rheumatoid Arthritis (RA) is a chronic, autoimmune disorder pri- arthritis includes steroidal drugs (corticosteroid), Non-Steroidal Anti-
marily affecting the synovial joints and damages the cartilage and Inflammatory Drugs (NSAIDs) (Ibuprofen and naproxen), Disease-
bones. RA annually affects about 0.51% of population in worldwide Modifying Anti-Rheumatic Drugs (DMARDs) (Methotrexate, lefluno-
and women are mostly affected than men at 3:1 ratio (Guo et al., mide). Even biological therapies such as infliximab (TNF-a blocking
2018; Silman and Pearson, 2002). RA is strongly associated to the auto- agents), rituximab (anti-CD 20 therapy) and abatacept are prescribed
immune response triggered by various genetic, epigenetic and some for RA treatment (Joshi and Dhaneshwar, 2014). Chemo and biological
environmental factors and its progression is classified into three stages therapies suppresses the inflammation and provide only temporary
as swelling with pain, inflamed synovium, finally bone and cartilage relief along with adverse side effects. Administration of Classic
destruction causing joint damage with pain (Smolen et al., 2018). Vari- DMARDs like methotrexate are reported with side effects like nausea,
ous proinflammatory cytokines (Interleukin-1b, 17, 34, TNF-a: vomiting, kidney malfunctioning, haematological adverse state includ-
Tumour Necrosis Factor), enzymes of Cyclooxygenase II (COX-II), Lysyl ing anemia and thrombocytopenia (Gowdie, 2011; Kumar et al., 2016).
Oxidase (LOX), Prostaglandin-Endoperoxide Synthase (PTGS), Reactive Herbal or natural medicines are an alternative treatment to the
Oxygen Species (ROS) of NO, H2O2, prostaglandins, Macrophage Colony harmful disease with lesser side effects (Murugesan et al., 2019;
Stimulating Factor (MCSF), Transforming Growth Factor (TGF) are Rengasamy et al., 2019). In India, there are more than 2500 plants
mainly involved in the RA pathology. Currently PTGS, COX and LOX species that are currently used as herbal medicaments (Pan et al.,
2013; Rengasamy et al., 2019). Further, many of the crude herbal-
* Corresponding author. based drugs and their constituents have a potent antioxidant activity
E-mail address: drsureshbiotech2003@gmail.com (S. Periyasamy). and scavenge free radicals, since these free radicals propagate

https://doi.org/10.1016/j.sajb.2019.12.019
0254-6299/© 2019 Published by Elsevier B.V. on behalf of SAAB.
46 S. Murugesan et al. / South African Journal of Botany 130 (2020) 4553

inflammatory responses and cause cartilage damage (Suroowan and 2.4. ABTS radical cation decolorization assay
Mahomoodally, 2018). Natural chemical constituents extracted from
medicinal plants can effectively interact and modulate the expression Free radical scavenging activity of Z. officinale was determined by
of pro-inflammatory signal on inflammation pathway, and reduces ABTS radical cation decolorization assay. Various concentrations of
the adverse arthritic effect (Amran et al., 2015). different ginger extract (20, 40, 60, 80 and 100 mg/mL) was added to
Zingiber officinale Rosc. (ginger) is a rhizomatous, monocotyledon- 3 mL of ABTS¢+ cation radical and incubated at room temperature for
ous plant that is extensively used around the world to treat a wide 30 min. Then, the sample was read at 734 nm and the percentage of
range of diseases such as rheumatic disorders, cold symptoms, fevers, inhibition was calculated using Eq. (2.1) (Jeyadevi et al., 2019).
gastrointestinal complications, motion sickness, bronchitis, and dia-
betes (Brahmbhatt et al., 2013). The biological and medicinal proper- 2.5. Nitric oxide radical scavenging assay
ties of Z. officinale were mainly attributed through its bioactive
constituents like 6-Gingerol, 6-shogaol, other phenolics and flavo- Aqueous solution of sodium nitroprusside spontaneously gener-
noids. Though NSAIDs and DMARDs are prescribed during chronic ates NO at physiological pH, which interacts with oxygen to produce
inflammation conditions, their long term usage ends up with gastro nitric oxide that can be measured through calorimetrical method.
intestinal disorders. Ginger being another potent source with rich 3 mL of reaction mixture containing sodium nitroprusside (10 mM in
secondary metabolites was proven to suppress the prostaglandin PBS: Phosphate buffer saline) and 1 mL of plant extracts were incu-
synthesis by inhibiting COX-I, COX-II, suppressing the leukotriene bated at 25 °C for 3 h. After incubation, 0.5 mL of reaction mixture
biosynthesis through 5-LOX inhibition and inhibiting the several was removed and 0.5 mL of Griess reagent (1% sulfanilamide, 5%
inflammatory cytokines expressions (Grzanna et al., 2005). In fact, H3PO4, 0.1% naphthalene diamine dihydrochloride) was added. The
the prime compound, 6-Gingerol was reported with various pharma- absorbance was read at 546 nm (Atere et al., 2018).
cological activities including anti-inflammatory, antioxidant, antican-
cer, anti-lipidemic, antidiabetic, analgesic and antipyretic properties 2.6. In vitro anti-arthritic activities
(Mahomoodally et al., 2019; Srinivasan, 2017). which can be utilized
in treating the arthritis and its associated inflammatory responses. Z. officinale extracts obtained from various organic solvents were
Considering the potential of ginger metabolites, the present study tested for the anti arthritic activities and results were compared with
was attempted to investigate the anti-rheumatic properties of Z. offi- standard anti-inflammatory drug (diclofenac).
cinale using in silico and in vitro approaches.
2.7. Protein denaturation inhibition assay
2. Materials and methods
Bovine serum albumin (5% of 2.4 mL) and 1 mL aqueous solution
2.1. Sample collection, authentication and extraction contains crude plant extracts were added in the reaction tube. The pH
was adjusted to 6.3 and incubated at 37 °C for 20 min. Then the sam-
The fresh rhizomes of ginger (Zingiber officinale Rosc.) were col- ples were heated at 57 °C for 30 min. After cooling the samples, 2.5 mL
lected from Ooty, Tamil Nadu, India and authenticated by Botanical PBS (pH 6.3) was added to each tube and made up to 5 mL. Turbidity
Survey of India (BSI) Coimbatore, Tamil Nadu, India. (BSI/ SRC/ 5/ 23/ was determined in spectrophotometer at 660 nm wavelength com-
2018/ TECH/ 2242). Shadow dried ginger was grided into powder and pared to control. The percentage inhibition of protein denaturation
used for the extraction (10 g of ginger powder in 150 mL of different was computed using Eq. (2.1) (Senthilkumar et al., 2017).
organic solvents). Using soxhlet apparatus the phytoconstituents
were extracted with different organic solvents (Methanol, Ethyl ace- 2.8. Membrane stabilization assay
tate, Hexane). Extracts were concentrated through rotary vacuum
evaporator at 45 °C for 10 mins (Yamato Scientific Co, Japan) The final To the assay reaction mixtures (4.5 mL) containing 1 mL of 0.15 M
concentrated extracts were stored in 20 °C and used for In vitro PBS (pH 7.4), 2 mL hypotonic saline solution (0.25% NaCl), and 1 mL
assays. aqueous solution containing Z. officinale extract, 0.5 mL human red
blood cell (HRBC) suspension [10% v/v] in normal saline was added.
For control, 1 mL of hypotonic saline was used instead of test solution
2.2. In vitro scavenging potential assays while red blood cells were absent in the control. The mixture was
incubated at 57 °C for 30 min. The tubes were chilled and then centri-
Antioxidant potential of different extracts of Z. officinale was fugation was carried out, finally absorbance of the supernatants was
determined using 2,2-diphenyl-1-picrylhydrazyl (DPPH), 2,20 -Azino- determined at 560 nm. Percentage of membrane stabilizing potential
bis-(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and Nitric oxide was calculated by Eq. (2.1) (Sharma et al., 2016).
(NO) activities and compared with standard butylated hydroxyto-
luene (BHT) 2.9. Proteinase inhibitory assay

2.3. DPPH free radical scavenging activity The reaction mixtures of 2 mL contained 0.06 mg trypsin, 1 mL of
25 mM tris-HCl buffers (pH 7.4) and 1 mL of Z. officinale extract. The
Different concentration of ginger extracts (20, 40, 60, 80 and mixtures were incubated at 37 °C for 10 min, and then 1 mL of 0.8%
100 mg/mL) were added to 3 mL of DPPH solution (0.135 mM). After of casein solution was added with 20 min incubation. Later, 2 mL of
20 min of incubation, the absorbance (Abs) was measured at 517 nm 70% of perchloric acid was added to stop the reaction proceeding. The
and compared with the control. DPPH radical scavenging activity was cloudy suspension was centrifuged and supernatant solution was
indicated by its decreasing absorbance at 517 nm with respective measured at 280 nm wavelength against buffer as blank. The percent-
concentrations (Jeyadevi et al., 2019). age of inhibition was calculated as per the Eq. (2.1) (Aziz, 2015).
ðAbs conAbs sampleÞ
% Inhibition activity ¼  100 ð2:1Þ 2.10. Gas Chromatography Mass Spectrometry (GC-MS) analysis
ðAbs conÞ
Where, Abs con is the absorbance of the control and Abs sample is The active phytochemicals responsible for the antioxidant and
the absorbance of the sample extract. anti inflammatory activity in the crude extract of Z. officinale was
S. Murugesan et al. / South African Journal of Botany 130 (2020) 4553 47

examined using gas chromatography mass spectrometry (GC 7890- bioavailability and mutagenicity were estimated using admetSAR
MS 5975C) Electron ionization mode with Triple-Axis Detector (Agi- server (Yang et al., 2019).
lent Technologies, Santa Clara, CA, USA). The capillary column of
DB5MS with internal diameter: 30 m £ 0.25 mm, film thickness: 2.14. Preparation of target RA proteins
0.25 mm was used for separation process. The sample injector tem-
perature was 250 °C in splitless mode and helium was used as a car- Crystalline structure of chief targets like COX-II (5F19), TNF-a
rier gar with the flow rate of 1 mL/min. The MS spectral data was (1THF), MCSF (5LXF), IL-1b (9ILB), MMP9 (1L6J) proteins were
obtained from the Compound Library database of NIST08s.LIB retrieved from RCBS-PDB (http://www.rcsb.org/pdb). Water mole-
(National Institute of Standards and Technology, US) based on the cules, ligands and undesired bonding were removed from crystalline
retention time and mass. structure of target proteins using Discovery Studio 2017R2. The miss-
ing hydrogen atoms, unfilled atom valency were ratified and energy
2.11. Computational studies minimization was achieved by force-field (Kumar et al., 2020).

The major GC-MS peak of bioactives was selected for further in silico 2.15. Molecular docking studies
analysis. The chief RA target proteins were selected from DrugBank
Database and Literature reports. Molecular docking analysis was per- Initially, the energy minimization process was performed to the 3D
formed on AutoDock (AD) inbuild vina PyRx 0.8 version software and structures of selected ligands using MMFF94 force field. The energy
the interaction studies were carried out on Discovery Studio 2017. minimized ligands (.sdf) and protein structures (.pdb) were converted
into PDBQT file format (.pdbqt). The protein-ligand docking studies
2.12. Preparation of ligands and analysis of drug likeliness were achieved by grid box approach for covering the binding sites. Eval-
uation of docking results was sorted via predicted binding energy
The crystal 3D structure of selected active compounds in metha- (-kcal/mol). A cluster analysis based on root mean square deviation val-
nolic extract of Z. officinale was obtained from PubChem Database. ues (RMSD values) and the lowest energy conformation of the most
Drug-likeliness properties including Topological polar surface area populated cluster was considered as the most trustable solution (Aru-
(TPSA), Atom Molar Refractivity (AMR), log P, Alog P, and physico- mugam et al., 2019).
chemical properties were analysed for the selected active compounds
using DruLiTo software (Lipinski et al., 2001). 2.16. Binding poses visualization and analysis of interactions

2.13. Pharmacokinetic and bioactivity properties The PyRx results were analysed using Discovery Studio 2017R2.
The different binding mode of ligands file obtained from the end of
The pharmacokinetic properties like human intestinal absorption the docking studies and that files were analysed on Discovery Studio
(HIA), water solubility, distribution (blood-brain barrier (BBB) perme- to check the best binding mode with the binding site cavity. The
ability log BS, central nervous system (CNS) permeability), metabo- protein-ligand interactions of H-bond, CH bond, Vander Waals
lism (Cytochrome P450 inhibitors), excretion (renal and total interaction, Alkyl, Pi-Alkyl, and Pi-Pi Alkyl interactions were viewed
clearance), and toxicity (hepatotoxicity, carcinogenicity) were evalu- and various atomic distance, angle were measured the same software
ated using the pkCSM web server (Pires et al., 2015). The oral (Arumugam et al., 2019; Kumar et al., 2020).

Fig. 1. In vitro Antioxidant and Anti arthritic activities of ZOME. (a) ABTS radical scavenging activity of different crude extracts of Zingiber officinale. (b) DPPH radical scavenging
activity of different extracts of Zingiber officinale. (c) NO radical scavenging activity of different extracts of Zingiber officinale. (d). Protein denaturation inhibition capacity of different
extracts of Zingiber officinale. (e) Anti proteinase effect of different extracts of Zingiber officinale. (f) Membrane stabilization properties of different extracts of Zingiber officinale.
48 S. Murugesan et al. / South African Journal of Botany 130 (2020) 4553

2.17. Statistical analysis ZOME exhibited 91.04§0.96% of inhibitory activity and least activity
exhibited by ZOHX (57.43§1.81%) (Fig 1. b). ZOME also showed the
The in vitro assay results were calculated as means § SD (n = 3). highest NO scavenging potential of 86.72§1.51% that was compara-
Data were analysed by comparing the different ginger extracts with ble with the activity of BHT (97.35§0.88%) (Fig 1. c).
appropriate standard. Protein denaturation mechanism plays a crucial role in inflamma-
tory pathway by increasing the auto antigen production in RA pathol-
3. Results ogy (Ludwig et al., 2017). NSAIDs like provides protective effect
against denaturation of protein (Saso et al., 2001). With respect to
3.1. In vitro studies the protein denaturation inhibitory assay using different ginger
extracts, ZOME showed significant inhibition with the higher inhibi-
ROS formation increases the oxidative stress that leads to autoim- tion capacity (81.68§1.66%) (Fig 1. d).
mune diseases including RA. The increasing lipid peroxidation Proteinase has been closely related to the arthritic mechanism
reduces the anti oxidant enzymes activities and decreases the Na⁺/ since it degrades the matrix tissues during the inflammation. Espe-
K⁺-ATPase functions. Increasing ROS level also damage the immuno- cially, MMPs are endopeptidases enzymes mainly involved in the car-
globulins, and affect the mechanism of DNA mismatch repair. From tilage matrix degradation and cause the destruction of bone joints
these previous reports, the ROS oxidative stress is an important path- (Itoh, 2017). The crude extract of Z. officinale contributed the anti-
ological role in RA progression (Di Dalmazi et al., 2016; Staron et al., proteinase activity, yet again the ZOME showed higher proteinase
2012). In our investigation, the potential antioxidant activities of inhibitory effect (82.72§1.48%) compared with Standard diclofenac
crude extract of Z. officinale was evaluated by DPPH, ABTS and NO (90.35§1.38%). In this particular assay, the inhibitory effect of the
assays, and the inhibitory effect was compared with the standard standard was higher when compared to the ZOME (Fig 1. e).
BHT. Among these extracts, methanolic extract showed the signifi- Lysis of lysosomes during inflammation releases harmful enzymes
cant activity in a dose dependent manner. Highest antiradical poten- in to the circulatory system. NSAIDs are used to suppress the release
tial was obtained from the ZOME at the maximum tested of lysosomal enzymes by neutralizing the membrane damage of lyso-
concentration (100 mg/mL). The percentage inhibition of ZOME was somes. Undesirable environmental conditions like hypotonic
found to be 86.26§0.97% in DPPH assay which was little lower when medium, over heat, and chemical medications containing Methotrex-
compared to the standard BHT activity (94.52§1.63%) and exhibited ate, hydroxychloroquine may affect the stability of RBC membrane
higher activity than the other extracts (Fig 1. a). Regarding the ABTS, and causes hemolysis (Held et al., 2018). Since RBCs mimics the

Fig. 2. The GCMS chromatogram of the methanolic extract of Zingiber officinale Rosc.
S. Murugesan et al. / South African Journal of Botany 130 (2020) 4553 49

Table 1 Table 3
Bioactive compounds derived from Zingiber officinale methanol Molecular docking of selected GCMS compounds with RA target proteins.
extract.
Compounds COX-2 IL1-Beta MCSF MMP-9 TNF Alpha
S. no RT % Peak Compound name
2,5-dibutylfuran 6.5 4.3 4.7 5.7 4.3
1 9.709 0.76 Decanal 6-Gingerol 7.8 5.7 5 6.7 4.7
2 13.417 7.76 Zingiberene 8-Gingerol 7.5 5.4 5.7 6.1 5.6
3 13.467 5.39 Curcumene Benzoic acid 6.1 4.6 4.9 6.2 4.8
4 13.534 2.47 Beta-bisabolene Dihydrocapsaicin 7.4 5.3 5.4 5.9 5.5
5 13.778 1.13 Germacrene D Dihydropseudoionone 4.4 3.2 3.6 4.9 3.4
6 13.861 5.02 Beta-sesquiphellandrene Ferulic acid ethyl ester 7.3 5.3 5.1 7.1 5.4
7 14.373 0.73 Nerolidol Geranylacetone 4.4 3.4 3.7 5 3.3
8 15.22 0.57 Alpha-terpineol Zingerone 6.9 5.2 5 7.4 5
9 15.506 0.54 Terpineol
10 16.017 0.55 Gamma-elemene
11 16.21 0.53 Beta-eudesmol secondary metabolites were identified (Fig 2). The polyphenol of 6-
12 16.252 0.85 Alpha-cedrene Gingerol the major peak (23.46%) on 23.25 min, followed by hexade-
13 16.537 2.07 Dihydropseudoionone
14 17.099 6.27 Zingerone
canoic acid (10.04%), zingiberene (7.76%), dihydrocapsaicin (7.37%),
15 17.242 0.61 Curcuphenol zingerone (6.27%), curcumene (5.39%), beta bisabolene (5.02%) octa-
16 18.005 2.32 Farnesol, acetate decanoic acid (4.78%) and 8-Gingerol (4.45%) (Table 1). The active
17 18.819 1.63 Geranylacetone compound, 6-Gingerol was reported for many potential biological
18 20.648 0.93 9,12-Octadecadienoic acid
properties including anti-cancer, antioxidant, anti-inflammatory and
19 21.386 1.32 Benzoic acid
20 22.519 0.81 3-Acetamidobenzoic acid inhibit the angiogenesis activities (Wang et al., 2014).
21 22.586 1.39 2,5-dibutylfuran
22 23.257 23.46 6-Gingerol
23 23.475 4.03 Ferulic acid ethyl ester
3.2. In silico studies
24 23.701 0.54 Carinol
25 24.255 10.22 Hexadecanoic acid
26 24.876 4.45 8-Gingerol
3.2.1. Drug-likeness properties
27 25.044 0.65 1,4-naphthoquinon The physiochemical properties of major 16 active compounds
28 25.798 4.78 Octadecanoic acid were analysed on DruLiTo. Around nine compounds obeyed the Lip-
29 26.428 7.37 Dihydrocapsaicin inski’s rule, especially, 6-Gingerol and 8-Gingerol showed higher
30 26.57 0.85 P-Toluic acid, pentadecyl ester
TPSA (66.76, 66.76) and AMR (80.8, 86.63) with suitable HBA, HBD
(Table 2) and nRB. TPSA and AMR are important physiochemical
lysosomal membrane, studying the protective effect of could be use- properties mainly involved in drug absorption, transport and pene-
ful to analyze the anti-inflammatory property. Here the crude extract tration mechanism (Ertl et al., 2000).
of ZOME expressed the greater stabilization effect (84.72§1.38%)
when compared to other extracts and also found to exhibit equiva- 3.2.2. Molecular docking studies
lent activity when compared with the BHT (93.35§1.33%) (Fig 1. f). The selected bioactives were docked with RA target proteins (COX-II,
Various biological studies have shown that many phenolic and fla- IL-1b, MCSF, MMP9 and TNF-a) using PyRx. Among these compounds,
vonoids significantly contributed to the antioxidant and anti-arthritic dihydrocapsaicin, 6-Gingerol and 8-Gingerol expressed the minimized
activities in herbal plants. Z. officinale, has abundant amount of poly- binding energy in kcal/mol (Table 3). COX-II with 6-Gingerol, and IL-1b
phenols like gingerol derivatives, shogaol, paradol and zingerone with 6-Gingerol showed the highest binding affinity (7.8, and
which might be responsible for the anti arthritic activities (Funk 5.7 kcal/mol). Similarly, growth factor molecule of MCSF with
et al., 2009; Semwal et al., 2015). 8-Gingerol and signaling molecule of TNF-a with 8-Gingerol exposed the
Since ZOME exhibited best antiradical and antiarthritic activities, potential binding affinity with minimized binding energy (5.7 and
it was further chosen for GCMS analysis, because it exhibited excel- 5.6 kcal/mol). Zingerone with endopeptidase of MMP9 complex exhib-
lent antiradical activities. Using GCMS results, 30 different ited the higher binding score with lesser binding energy (7.4 kcal/mol).

Table 2
Physicochemical properties of the active compounds and accordance with the rules of drug-likeness.

Ligand Compounds MW log P Alog P HBA HBD TPSA AMR nRB

1 2,5-Dibutylfuran 180.15 3.963 0.65 1 0 9.23 53.26 6


2 Hexadecanoic acid 256.24 7.57 3.898 2 1 37.3 55.52 14
3 6-Gingerol 294.18 2.437 1.597 4 2 66.76 80.8 10
4 8-Gingerol 322.21 3.575 2.173 4 2 66.76 86.63 12
5 Benzoic acid 122.04 0.982 0.968 2 1 37.3 36.96 1
6 Beta-Sesquiphellandrene 204.19 6.399 3.192 0 0 0 68.91 4
7 Bisabolene 204.19 5.551 3.375 0 0 0 69.93 4
8 Curcumene 202.17 5.761 3.855 0 0 0 71.56 4
9 Dihydrocapsaicin 307.21 4.503 0.766 4 2 58.56 85.05 11
10 Dihydropseudoionone 194.17 3.618 2.825 1 0 17.07 63.79 6
11 Farnesol acetate 264.21 5.086 3.951 2 0 26.3 84.37 9
12 Ferulic acid ethyl ester 222.09 1.524 0.769 4 1 55.76 64.51 5
13 Geranylacetone 194.17 3.619 2.825 1 0 17.07 63.79 6
14 Germacrene D 204.19 6.294 2.797 0 0 0 69.55 1
15 Zingerone 194.09 0.761 0.011 3 1 46.53 58.17 4
16 Zingiberene 204.19 6.354 3.339 0 0 0 70.13 4
50 S. Murugesan et al. / South African Journal of Botany 130 (2020) 4553

Table 4
Interacting residues of RA targets with selected bioactive compounds from Z. officinale.

Compounds COX-II IL-1b MCSF MMP9 TNF-a

6-Gingerol TRP387, HIS388, LEU391, ASN7, GLU64, LEU62, LYS65, ARG66, PHE67, MET65, ARG51, THR96, LEU39, ILE154, SER9, ILE155, VAL13
HIS386, TYR385 SER153, LYS63 PRO110, LEU113, LEU114 LEU44
8-Gingerol ASN382, HIS386, HIS207, SER125, LEU134, ASP142, GLN58, ASP59, LEU55, LYS88 ARG51, TRY52, ASN38, TYR59, PRO8, ILE154, THR7,
VAL295, ILE408, LEU391, PRO131, PHE133 ASP182, LYS184, ARG95, VAL13, LEU36, ILE155
TYR404 LEU39
Zingerone ALA199, HIS386, ALA202, SER43, LEU62, PRO91 TYR49, LYS116 HIS401, LEU397, LEU418 ARG6, LEU36, PRO8, VAL13
GLN203

3.2.3. Interaction studies important properties for oral drug designing process. All the com-
The rigid docking results were visualized using Discovery studio pounds showed above 90% of intestinal absorption value with suit-
for analysis of interaction. The best binding poses of protein-small able aqueous solubility (8-Gingerol: 91.716; 6-Gingerol: 92.416, and
molecule interactions were visualized and tabulated in Table 4. The zingerone: 94.103). The in silico analysis of compound distribution
strongest interaction was observed in the 8-Gingerol with RA targets (log BB; log PS), metabolism (cytochrome P450 inhibitors) excretion
protein complexes. COX-II with 8-Gingerol complex formed one H- (total clearance and renal clearance) and toxicity (oral toxicity, hepa-
 
bond (ASN382; 1.95 A), two Pi-Pi stacked interaction (HIS386; 4.18 A, totoxicity, AMES toxicity) were analysed and tabulated in Table 5. All
 
HIS207; 4.34 A), three alkyl interaction (VAL295; 4.61 A, ILE408; the compounds did not show any toxicity including AMES, oral acute
  
4.79 A, LEU391; 5.43 A) and one Pi-Alkyl interaction (TYR404; 5.13 A) and hepatotoxicity. Carcinogenicity and mutagenicity have not been
(Fig 3). The protein-small molecule complex of 8-Gingerol-MCSF observed in any compounds from the in silico prediction. Overall 8-

showed one hydrogen bonding (GLN58; 3.38 A), one CH bonding Gingerol showed higher oral availability as predicted by admetSAR.

(ASP59; 3.3 A) and two vander waals attraction (LEU55, LYS88). The The dosage of maximum tolerance was predicted for all three com-
complex of 8-Gingerol and MMP9 exhibited the three covalent bonds pounds, 8-Gingerol (0.694), 6-Gingerol (0.635), and zingerone
  
(ARG51; 3.20 A, TRY52; 2.89 A, ASN38; 2.72 A), two CH bond (0.544). Using Molinspiration Cheminformatics web server, all
 
(ASP182; 3.59 A, LYS184; 3.52 A) and two hydrophobic interactions selected compounds relative bioactivity scores were predicted and
 
(ARG95; 3.76 A, LEU39; 4.69 A). The receptor-ligand complex of TNF- summarized in Table 5. The highest score indicates better biological
a with 8-Gingerol

significantly interacted with three covalent bonds
 
activities. In an over view, 8-Gingerol and 6-Gingerol showed better
(TYR59; 2.89 A, PRO8; 2.99 A, ILE154; 2.11 A), one carbon hydrogen bioactivity score and acts as an enzyme inhibitor (0.34 and 0.38).
 
bond (THR7; 3.36 A), one Pi-Sigma interaction (VAL13; 3.92 A) and
 
two Pi-Alkyl interaction (LEU36; 5.06 A, ILE155; 4.98 A). 8-Gingerol
 4. Discussion
with IL-1b complex formed the three covalent bond (SER125; 2.88 A,
 
LEU134; 2.40 A, ASP142; 2.A), one CH bond (PHE133) and three
RA being a chronic autoimmune disorder is manifested with red-
hydrophobic interactions (PRO131, PHE133).
dish joints, swelling, and fatigue along with increased mortality rate.
6-Gingerol with RA target protein complexes also shown notable
Initially, the synovial fluid lining the joints is majorly affected fol-
strongest interaction. Especially, protein-ligand complex of 6-Gingerol
 lowed by cartilage and bone damages which may even worsen up to
with IL-1b formed the 5 H-bond interactions (ASN7; 1.98 A , GLU64;
    multi organ failure. Women are at the higher risk end with the arthri-
2.79 A, LEU62; 2.63 A, LYS65; 2.07 A, SER153; 1.96 A) and one Pi-Alkyl
 tis incidence along with other striking causes including genetic fac-
interaction (LYS63; 4.75 A) (Fig 3). The polyphenolic compound Zinger-
tors, stress, smoking and viral infections (Mcinnes and Schett, 2011)
one had shown significant interaction to the RA targets with suitable
The aforementioned factors influence the post transcriptional alter-
covalent and hydrophobic interactions shown in Table 4.
ations and it releases the citrullinated proteins, rheumatoid factors and
other self-proteins like vimentin, a-enolase, histones, fibronectin,
3.2.4. Pharmacokinetic and bioactivity properties fibrinogen, and collagen (Kurowska et al., 2017). These proteins are
From the docking and visualization studies, 8-Gingerol, 6- not considered as self proteins, so Anti citrullinated protein antibodies
Gingerol and Zingerone were selected for pharmacokinetic and bio- (ACPAs) are produced in response to it. Several inter and intra cellular
activity analysis. The water solubility and intestinal absorption are signaling mechanisms are involved in the formation of immune

Fig. 3. Schematic diagrams of RA targets with 8-gingerol complex interaction using Discovery studio. (a) IL-1b with 8-Gingerol. (b) MMP9 with 8-Gingerol.
S. Murugesan et al. / South African Journal of Botany 130 (2020) 4553 51

Table 5
Pharmacokinetic and bioactivity properties of selected active compounds from Z. officinale.

ADMET Properties name Compound names

Zingerone 6-Gingerol 8-Gingerol

Bioavailability Human oral bioavailability 0.7857 0.7714 0.8000


Absorption Water solubility (log S mol/L) 1.7 3.164 3.795
Intestinal absorption (%) 94.103 92.416 91.716
Distribution BBB permeability (log BB) 0.006 0.727 0.794
CNS permeability (log PS) 2.175 2.788 2.799
Metabolism CYP2D6 substrate (Yes/No) No No No
CYP3A4 substrate (Yes/No) No No Yes
Excretion Total Clearance (log ml/min/kg) 0.307 1.339 1.4
Renal OCT2 substrate (Yes/No) No No No
Toxicity AMES toxicity (Yes/No) No No No
Max. tolerated dose human (mg/kg/day) 0.544 0.635 0.694
Oral Rat Acute Toxicity LD50 (mol/kg) 2.129 1.958 1.957
Hepatotoxicity (Yes/No) Yes No No
Carcinogenicity (Yes/No) No No No
Mutagenicity (Yes/No) No No No
Bioactivity score GPCR ligand 0.58 0.16 0.18
Ion channel modulator 0.18 0.04 0.03
Kinase inhibitor 1.15 0.33 0.27
Nuclear receptor ligand 0.59 0.2 0.23
Protease inhibitor 0.72 0.15 0.2
Enzyme inhibitor 0.07 0.38 0.34

Fig. 4. Rheumatoid arthritis progression mechanism and pathology: RANKL- Receptor activator of nuclear factor kappa-B ligand; TGF-b- Transforming growth factor beta; MMPs-
Matrix metalloproteinases; M-CSF- Macrophage Colony-Stimulating Factor; GM-CSF- Granulocyte Monocyte Colony-Stimulating Factor COX-II- Cyclooxygenase-2; IL17- Interleukin
17; IL34- Interleukin 34; Th1- T helper type 1, Th17- T helper type 17.

complex. Mast cells, macrophages and natural killer cells are the vital Nearly 60% of people look for an alternative herbal remedy for the
players in the pathology of RA progression. TNF-a, IL-1, IL-6, IL-12, IL- long term cure of RA (Rao et al., 1999) Chopra et al., 2000 demon-
15, IL-18, and IL-23 are few cytokines secreted by macrophages, espe- strated the antiarthritic potential of an ayurvedic tablet, RA-1 contain-
cially TNF-a and IL-6 regulates the cytokines and chemokines activa- ing of Boswellia serrata, Withania somnifera, Z. officinale and Curcuma
tion that aggravate inflammatory reaction and causes cartilage longa in 182 patients for 4 months. From the increase in the daily dose
damage (Smolen et al., 2018) (Fig 4). Nowadays, extra cellular specific of 444 mg, almost 50% of arthritic patients showed significantly
biological DMARDs like sarilumab and adalimumab are available to reduced the swelling of bones when compared with the placebo
reduce the inflammation. However, NSAIDs simply acts as timely pain patients. Studies using herbal medicines still need improvised and
subsidizers rather having a long term mechanistic modification in pre- standardized trials considering the efficacy and safety for the global
venting the joint and bone damage (Rein and Mueller, 2017). recognition of traditional herbal medicines (Chopra et al., 2000).
52 S. Murugesan et al. / South African Journal of Botany 130 (2020) 4553

Our investigation majorly focused on searching of nutraceutically Chopra, A., Lavin, P., Patwardhan, B., Chitre, D., 2000. Randomized double blind trial of
valuable new molecules from herbal plant of Z. officinale with suitable an Ayurvedic plant derived formulation for treatment of rheumatoid arthritis.
Journal of Rheumatology 27, 1365–1372.
biological efficacy and least toxicity for treatment of arthritis. The Di Dalmazi, G., Hirshberg, J., Lyle, D., Freij, J.B., Caturegli, P., 2016. Reactive oxygen spe-
present study has revealed that the antiradical and anti-arthritic cies in organ-specific autoimmunity. Autoimmunity Highlights 7, 11. https://doi.
activity of methanolic extract of the Z. officinale through in vitro and org/10.1007/s13317-016-0083-0.
El-Nabarawy, M.A., El-Kafafi, S.H., Hamza, M.A., Omar, M.A., 2015. The effect of some
in silico studies. From this result, 8-Gingerol is specifically chosen for factors on stimulating the growth and production of active substances in Zingiber
the further study. 6-Gingerol, 8-Gingerol and 10-Gingerol are few officinale callus cultures. Annals of Agricultural Sciences 60, 1–9. https://doi.org/
non-volatile, pungent, lipophilic compounds of ginger rhizome, 10.1016/j.aoas.2014.11.020.
Ertl, P., Rohde, B., Selzer, P., 2000. Fast calculation of molecular polar surface area as a
which are reported as DPPH, super oxide and hydroxyl radical scav-
sum of fragment-based contributions and its application to the prediction of drug
enging agents (Baliga et al., 2015). Other active ingredients in ginger transport properties. Journal of Medicinal Chemistry 43, 3714–3717. https://doi.
including paradol, shogaol showed strong inhibition of COX-II org/10.1021/jm000942e.
Funk, J.L., Frye, J.B., Oyarzo, J.N., Timmermann, B.N., 2009. Comparative effects of two gin-
enzyme that is involved in inflammatory responses (Van Breemen
gerol-containing Zingiber officinale extracts on experimental rheumatoid arthritis.
et al., 2011). No such proof is available to substantiate the anti Journal of Natural Products 72, 403–407. https://doi.org/10.1021/np8006183.
arthritic mechanistic role of 8-Gingerol. Owing to the increasing Gowdie, P., 2011. https://www.who.int/selection_medicines/committees/expert/18/
demand for the gingerol content in the medical care, 8-Gingerol applications/Child_DMARD.pdf.
Grzanna, R., Lindmark, L., Frondoza, C.G., 2005. Gingeran herbal medicinal product
content is being mass cultured using callus suspension technique with broad anti-inflammatory actions. Journal of Medicinal Food 8, 125–132.
using different elicitors and precursors that may intervene with the https://doi.org/10.1089/jmf.2005.8.125.
parent phenylpropanoid pathway involved in gingerol production Guo, Q., Wang, Y., Xu, D., Nossent, J., Pavlos, N.J., Xu, J., 2018. Rheumatoid arthritis:
pathological mechanisms and modern pharmacologic therapies. Bone Research 6,
(El-Nabarawy et al., 2015). 15. https://doi.org/10.1038/s41413-018-0016-9.
Held, J., Mosheimer-feistritzer, B., Gruber, J., Mur, E., Weiss, G., 2018. Methotrexate
therapy impacts on red cell distribution width and its predictive value for cardio-
5. Conclusion
vascular events in patients with rheumatoid. arthritis 2, 1–7. https://doi.org/
10.1186/s41927-018-0012-0.
The present study demonstrated that the methanolic extract of Itoh, Y., 2017. Metalloproteinases in rheumatoid arthritis: potential therapeutic targets
Zingiber officinale favourably prevented the ROS damage and sup- to improve current therapies. Progress in Molecular Biology and Translational Sci-
ence 148, 327–338. https://doi.org/10.1016/bs.pmbts.2017.03.002.
presses the inflammatory responses. GC-MS analysis revealed the Jeyadevi, R., Ananth, D.A., Sivasudha, T., 2019. Hepatoprotective and antioxidant activity
presence of 30 bioactives. The selected bioactive compounds upon of Ipomoea staphylina. linn 5, 18. https://doi.org/10.1186/s40816-019-0112-4.
docking with arthritic targets revealed that 8-Gingerol, 6-Gingerol Joshi, P., Dhaneshwar, S.S., 2014. An update on disease modifying antirheumatic drugs.
Inflammation & Allergy - Drug Targets 13, 249–261.
and Zingerone as the best anti-arthritic compounds. With further Kumar, L.D., Karthik, R., Gayathri, N., Sivasudha, T., 2016. Advancement in contempo-
screening, 8-Gingerol was selected as the chief arthritic compound of rary diagnostic and therapeutic approaches for rheumatoid arthritis. Biomedicine
interest that is being produced in Zingiber officinale through the callus & Pharmacotherapy = Biomedecine & Pharmacotherapie 79, 52–61. https://doi.
org/10.1016/j.biopha.2016.02.001.
culture techniques using elicitors to adopt a cost effective technique Kumar, L.D., Prathiviraj, R., Selvakumar, M., Guna, R., Abbirami, E., Sivasudha, T., 2020.
in understanding the mechanism of action of 8-Gingerol in both the HRLC-ESI-MS based identification of active small molecules from cissus quadrangula-
cell line and animal models. ris and likelihood of their action towards the primary targets of osteoarthritis. Journal
of Molecular Structure https://doi.org/10.1016/j.molstruc.2019.127048.
Kurowska, W., Kuca-Warnawin, E.H., Radzikowska, A., Maslinski, W., 2017. The role of
Declaration of competing interest anti-citrullinated protein antibodies (ACPA) in the pathogenesis of rheumatoid
arthritis. Central Journal of Immunology 42, 390–398. https://doi.org/10.5114/
ceji.2017.72807.
All authors declared that there are no conflicts of interest Lipinski, C.A., Lombardo, F., Dominy, B.W., Feeney, P.J., 2001. Experimental and compu-
tational approaches to estimate solubility and permeability in drug discovery and
development settings. Advanced Drug Delivery Reviews 46, 3–26.
Acknowledgments Ludwig, R.J., Vanhoorelbeke, K., Leypoldt, F., Kaya, Z., Bieber, K., McLachlan, S.M., et al.,
2017. Mechanisms of autoantibody-induced pathology. Frontiers in Immunology
We acknowledge Department of Science and Technology-Science 8, 603. https://doi.org/10.3389/fimmu.2017.00603.
Mahomoodally, M.F., Aumeeruddy, M.Z., Rengasamy, K.R.R., Roshan, S., Hammad, S.,
and Engineering Research Board (DST-SERB) New Delhi, India for pro-
Pandohee, J., Hu, X., et al., 2019. Ginger and its active compounds in cancer ther-
viding the financial support to conduct this study (Project no.: EEQ/ apy: from folk uses to nano-therapeutic applications. Seminars in Cancer Biology
2017/000447). https://doi.org/10.1016/j.semcancer.2019.08.009.
Mcinnes, I.B., Schett, G., 2011. The pathogenesis of rheumatoid arthritis New England.
Journal of Medicine 365 (23), 2205–2219.
Reference Murugesan, S., Kumar Lakshmanan, D., Arumugam, V., Alexander, R.A., 2019. Nutri-
tional and therapeutic benefits of medicinal plant pithecellobium dulce (Faba-
Amran, A.Z., Jantan, I., Dianita, R., Buang, F., 2015. Protective effects of the standardized ceae): a review article info. Journal of Applied Pharmaceutical Science 9, 130–139.
extract of Zingiber officinale on myocardium against isoproterenol-induced bio- https://doi.org/10.7324/JAPS.2019.90718.
chemical and histopathological alterations in rats. Pharmaceutical Biology 53, Pan, S., Zhou, S., Gao, S., Yu, Z., Zhang, S., Tang, M., et al., 2013. New Perspectives on
1795–1802. https://doi.org/10.3109/13880209.2015.1008147. How to Discover Drugs from Herbal Medicines: CAM’s Outstanding Contribution
Arumugam, V., Venkatesan, M., Murugesan, S., Ayyasamy, R., Ramachandran, S., to Modern Therapeutics. Evidence-Based Complementary and Alternative Medi-
Sundaresan, U., et al., 2019. Metabolite profiling, molecular docking and in vitro cine 125. https://doi.org/10.1155/2013/627375.
anticancer potential of marine ascidian Didemnum sp. Process Biochemistry (Bark- Pires, D.E.V, Blundell, T.L., Ascher, D.B., 2015. pkCSM: predicting small-molecule pharma-
ing, UK). https://doi.org/10.1016/J.PROCBIO.2019.09.021. cokinetic and toxicity properties using graph-based signatures. Journal of Medicinal
Atere, T.G., Akinloye, O.A., Ugbaja, R.N., Ojo, D.A., Dealtry, G., 2018. In vitro antioxidant Chemistry 58, 4066–4072. https://doi.org/10.1021/acs.jmedchem.5b00104.
capacity and free radical scavenging evaluation of standardized extract of Costus Rao, J.K., Mihaliak, K., Kroenke, K., Bradley, J., Tierney, W.M., Weinberger, M., 1999. Use
afer leaf. Food Science and Human Wellness 7, 266–272. https://doi.org/10.1016/j. of complementary therapies for arthritis among patients of rheumatologists.
fshw.2018.09.004. Annals of Internal Medicine 131, 409–416. https://doi.org/10.7326/0003-4819-
Aziz, M.A., 2015. Qualitative phytochemical screening and evaluation of anti-inflam- 131-6-199909210-00003.
matory, analgesic and antipyretic activities of Microcos paniculata barks and fruits. Rein, P., Mueller, R.B., 2017. Treatment with biologicals in rheumatoid arthritis: an
Journal of Integrative Medicine 13, 173–184. https://doi.org/10.1016/S2095-4964 overview. Rheumatology and Therapy 4, 247–261. https://doi.org/10.1007/
(15)60179-0. s40744-017-0073-3.
Baliga, M.S., Latheef, L., Haniadka, R., Fazal, F., Mane, P.P., Kalekhan, F., et al., 2015. Gin- Rengasamy, K.R.R., Khan, H., Gowrishankar, S., Lagoa, R.J.L., Mahomoodally, F.M.,
ger (Zingiber officinale roscoe) in the treatment of osteoarthritis: clinical observa- Khan, Z., et al., 2019. The role of flavonoids in autoimmune diseases: therapeutic
tions and mechanistic insights. Foods and Dietary Supplements in the Prevention updates. Pharmacology & Therapeutics 194, 107–131. https://doi.org/10.1016/j.
and Treatment of Disease in Older Adults. Elsevier Inc https://doi.org/10.1016/ pharmthera.2018.09.009.
B978-0-12-418680-4.00012-9. Saso, L., Valentini, G., Casini, M.L., Grippa, E., Gatto, M.T., Leone, M.G., et al., 2001. Inhi-
Brahmbhatt, M., Gundala, S.R., Asif, G., Shamsi, S.A., Aneja, R., 2013. Ginger phytochem- bition of heat-induced denaturation of albumin by nonsteroidal antiinflammatory
icals exhibit synergy to inhibit prostate cancer cell proliferation. Nutrition and drugs (NSAIDs): pharmacological implications. Archives of Pharmacal Research 24,
Cancer 65, 263–272. https://doi.org/10.1080/01635581.2013.749925. 150–158. https://doi.org/10.1007/bf02976483.
S. Murugesan et al. / South African Journal of Botany 130 (2020) 4553 53

Semwal, R.B., Semwal, D.K., Combrinck, S., Viljoen, A.M., 2015. Gingerols and shogaols: Srinivasan, K., 2017. Ginger rhizomes (Zingiber officinale): a spice with multiple health
important nutraceutical principles from ginger. Phytochemistry 117, 554–568. beneficial potentials. Pharma Nutrition 5, 18–28. https://doi.org/10.1016/j.
https://doi.org/10.1016/j.phytochem.2015.07.012. phanu.2017.01.001.
Senthilkumar, N., Nandhakumar, E., Priya, P., Soni, D., Vimalan, M., Vetha Potheher, I., 2017. Staron , A., Makosa,
˛ G., Koter-Michalak, M., 2012. Oxidative stress in erythrocytes from
Synthesis of ZnO nanoparticles using leaf extract of tectona grandis (L.) and their anti- patients with rheumatoid arthritis. Rheumatology International 32, 331–334.
bacterial{,} anti-arthritic{,} anti-oxidant and in vitro cytotoxicity activities. New Jour- https://doi.org/10.1007/s00296-010-1611-2.
nal of Chemistry 41, 10347–10356. https://doi.org/10.1039/C7NJ02664A. Suroowan, S., Mahomoodally, F., 2018. Herbal products for common auto-inflamma-
Sharma, A., Goyal, R., Sharma, L., 2016. Potential biological efficacy of Pinus plant spe- tory disorders - Novel Approaches. Combinatorial Chemistry & High Throughput
cies against oxidative, inflammatory and microbial disorders. BMC Complementary Screening 21, 161–174. https://doi.org/10.2174/1386207321666180213093449.
and Alternative Medicine (Electronic Resource) 16, 35. https://doi.org/10.1186/ van Breemen, R.B., Tao, Y., Li, W., 2011. Cyclooxygenase-2 inhibitors in ginger (Zingiber
s12906-016-1011-6. officinale). Fitoterapia 82, 38–43. https://doi.org/10.1016/j.fitote.2010.09.004.
Silman, A.J., Pearson, J.E., 2002. Epidemiology and genetics of rheumatoid arthritis. Wang, S., Zhang, C., Yang, G., Yang, Y., 2014. Biological properties of 6-gingerol: a brief
Arthritis Research 4 (Suppl 3), S265–S272. https://doi.org/10.1186/ar578. review. Natural Products Communications 9, 1027–1030.
Smolen, J.S., Aletaha, D., Barton, A., Burmester, G.R., Emery, P., Firestein, G.S., et al., Yang, H., Lou, C., Sun, L., Li, J., Cai, Y., Wang, Z., et al., 2019. admetSAR 2.0: web-service
2018. Rheumatoid arthritis. Nature Reviews Disease Primers 4, 18001. https://doi. for prediction and optimization of chemical ADMET properties. Bioinformatics 35,
org/10.1038/nrdp.2018.1. 1067–1069. https://doi.org/10.1093/bioinformatics/bty707.

You might also like